Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 28
1.
Ann Biomed Eng ; 2023 Jul 12.
Article En | MEDLINE | ID: mdl-37436565

Tissue-engineering and cell-based strategies provide an intriguing approach to treat complex conditions such as those of the endocrine system. We have previously developed a cell-based hormone therapy (cHT) to address hormonal insufficiency associated with the loss of ovarian function. To assess how the cHT strategy may achieve its efficacy, we developed a mathematical model to determine if known autocrine, paracrine, and endocrine effects of the native hypothalamus-pituitary-ovary (HPO) axis could explain our previously observed effects in ovariectomized rats following treatment with cHT. Our model suggests that cHT constructs participate in the complex machinery of the HPO axis. We were able to describe the in vivo behaviors of estrogen, progesterone, follicle-stimulating hormone (FSH), luteinizing hormone (LH), inhibin, and androgen with good accuracy. A sensitivity analysis indicated that some parameters impact the broader HPO system more than others, but that most changes in model parameters led to proportional changes in the system. We also conducted a predictive analysis on the effect of cHT dose on HPO axis hormones and found that, with the exception of estrogen, the other HPO hormones analyzed reach a saturation level within the physically possible number of constructs.

2.
Cell Tissue Res ; 386(1): 145-156, 2021 Oct.
Article En | MEDLINE | ID: mdl-34415395

Alternative methods to obtain mature oocytes are still needed for women with premature ovarian failure (POF). Oogonial stem cells (OSCs), found in adult ovaries, have provided insight into potential paths to treating infertility. Previously, the DDX4 antibody marker alone was utilized to isolate OSCs; however, extensive debate over its location in OSCs versus resulting oocytes (transmembrane or intracytoplasmic) has raised doubt about the identity of these cells. Separate groups, however, have efficiently isolated OSCs using another antibody marker Ifitm3 which is consistently recognized to be transmembrane in location. We hypothesized that by using anti-DDX4 and anti-IFITM3 antibodies, in combination, with MACS, we would improve the yield of isolated OSCs versus using anti-DDX4 antibodies alone. Our study supports earlier findings of OSCs in ovaries during the entire female lifespan: from reproductive age through post-menopausal age. MACS sorting ovarian cells using a the two-marker combination yielded a ~ twofold higher percentage of OSCs from a given mass of ovarian tissue compared to existing single marker methods while minimizing the debate surrounding germline marker selection. During in vitro culture, isolated cells retained the germline phenotype expression of DDX4 and IFITM3 as confirmed by gene expression analysis, demonstrated characteristic germline stem cell self-assembly into embryoid bodies, and formed > 40 µm "oocyte-like" structures that expressed the early oocyte markers GDF9, DAZL, and ZP1. This enhanced and novel method is clinically significant as it could be utilized in the future to more efficiently produce mature, secondary oocytes, for use with IVF/ICSI to treat POF patients.


Biomedical Research/methods , Fertility/physiology , Oogonial Stem Cells/metabolism , Female , Humans
3.
Reprod Sci ; 28(6): 1573-1595, 2021 06.
Article En | MEDLINE | ID: mdl-33877644

Diseases, disorders, and dysfunctions of the female reproductive tract tissues can result in either infertility and/or hormonal imbalance. Current treatment options are limited and often do not result in tissue function restoration, requiring alternative therapeutic approaches. Regenerative medicine offers potential new therapies through the bioengineering of female reproductive tissues. This review focuses on some of the current technologies that could address the restoration of functional female reproductive tissues, including the use of stem cells, biomaterial scaffolds, bio-printing, and bio-fabrication of tissues or organoids. The use of these approaches could also be used to address issues in infertility. Strategies such as cell-based hormone replacement therapy could provide a more natural means of restoring normal ovarian physiology. Engineering of reproductive tissues and organs could serve as a powerful tool for correcting developmental anomalies. Organ-on-a-chip technologies could be used to perform drug screening for personalized medicine approaches and scientific investigations of the complex physiological interactions between the female reproductive tissues and other organ systems. While some of these technologies have already been developed, others have not been translated for clinical application. The continuous evolution of biomaterials and techniques, advances in bioprinting, along with emerging ideas for new approaches, shows a promising future for treating female reproductive tract-related disorders and dysfunctions.


Genital Diseases, Female/therapy , Genitalia, Female , Regenerative Medicine/methods , Tissue Engineering/methods , Adult Stem Cells , Cell Culture Techniques, Three Dimensional , Embryonic Stem Cells , Fallopian Tubes , Female , Genital Diseases, Female/physiopathology , Genitalia, Female/physiopathology , Gonadal Steroid Hormones , Humans , In Vitro Oocyte Maturation Techniques , Oogenesis , Ovary , Pregnancy , Uterus
4.
Ann Biomed Eng ; 48(3): 1058-1070, 2020 Mar.
Article En | MEDLINE | ID: mdl-31367915

Loss of ovarian function (e.g., due to menopause) leads to profound physiological effects in women including changes in sexual function and osteoporosis. Hormone therapies are a known solution, but their use has significantly decreased due to concerns over cardiovascular disease and certain cancers. We recently reported a tissue-engineering strategy for cell hormone therapy (cHT) in which granulosa cells and theca cells are encapsulated to mimic native ovarian follicles. cHT improved physiological outcomes and safety compared to pharmacological hormone therapies in a rat ovariectomy model. However, cHT did not achieve estrogen levels as high as ovary-intact animals. In this report, we examined if hormone secretion from cHT constructs is impacted by incorporation of bone marrow-derived mesenchymal stem cells (BMSC) since these cells contain regulatory factors such as aromatase necessary for estrogen production. Incorporation of BMSCs led to enhanced estrogen secretion in vitro. Moreover, cHT constructs with BMSCs achieved estrogen secretion levels significantly greater than constructs without BMSCs in ovariectomized rats from 70 to 90 days after implantation, while also regulating pituitary hormones. cHT constructs with BMSC ameliorated estrogen deficiency-induced uterine atrophy without hyperplasia. The results indicate that inclusion of BMSC in cHT strategies can improve performance.


Cell- and Tissue-Based Therapy , Estrogens/metabolism , Granulosa Cells/metabolism , Hormone Replacement Therapy , Mesenchymal Stem Cells/metabolism , Theca Cells/metabolism , Tissue Engineering , Animals , Aromatase/metabolism , Cells, Cultured , Female , Follicle Stimulating Hormone/metabolism , Luteinizing Hormone/metabolism , Ovariectomy , Progesterone/metabolism , Rats, Inbred F344 , Uterus/anatomy & histology
5.
Prostate ; 79(9): 1053-1058, 2019 06.
Article En | MEDLINE | ID: mdl-30958903

Protein Kinase D1 (PrKD1) functions as a tumor and metastasis suppressor in several human cancers by influencing cell-cycle progression. However, the exact mechanism of cell-cycle regulation by PrKD1 is unclear. Overexpression and ectopic expression of PrKD1 induces G1 arrest in cancer cell lines. Because checkpoint kinases (CHEKs) are known to play a role in progression through the G1 phase, we downregulated CHEK1, which did not overcome the G1 arrest induced by PrKD1. Using in vitro phosphorylation and Western blot assays, we showed that PrKD1 phosphorylates all CDC25 isoforms (known substrates of CHEK kinases), independent from CHEK kinases, suggesting that direct phosphorylation of CDC25 by PrKD1 may be an alternate mechanism of G1 arrest. The study has identified a molecular mechanism for the influence of PrKD1 in cell-cycle progression.


Checkpoint Kinase 1/metabolism , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/pathology , Protein Kinase C/metabolism , cdc25 Phosphatases/metabolism , Cell Line, Tumor , Down-Regulation , G1 Phase Cell Cycle Checkpoints , Humans , Male , Phosphorylation , Prostatic Neoplasms/metabolism
6.
Nat Commun ; 8(1): 1858, 2017 12 05.
Article En | MEDLINE | ID: mdl-29208899

Safe clinical hormone replacement (HR) will likely become increasingly important in the growing populations of aged women and cancer patients undergoing treatments that ablate the ovaries. Cell-based HRT (cHRT) is an alternative approach that may allow certain physiological outcomes to be achieved with lower circulating hormone levels than pharmacological means due to participation of cells in the hypothalamus-pituitary-ovary feedback control loop. Here we describe the in vivo performance of 3D bioengineered ovarian constructs that recapitulate native cell-cell interactions between ovarian granulosa and theca cells as an approach to cHRT. The constructs are fabricated using either Ca++ or Sr++ to crosslink alginate. Following implantation in ovariectomized (ovx) rats, the Sr++-cross-linked constructs achieve stable secretion of hormones during 90 days of study. Further, we show these constructs with isogeneic cells to be effective in ameliorating adverse effects of hormone deficiency, including bone health, uterine health, and body composition in this rat model.


Granulosa Cells/transplantation , Hormone Replacement Therapy/methods , Ovary/cytology , Alginates/chemistry , Animals , Bone Density , Calcium/chemistry , Cell Transplantation/methods , Estrogens/metabolism , Female , Hormones/blood , Osteocalcin/blood , Primary Ovarian Insufficiency/therapy , Rats, Inbred F344 , Strontium/chemistry , Theca Cells/transplantation , Uterus/physiology
7.
Oncotarget ; 8(45): 78811-78824, 2017 Oct 03.
Article En | MEDLINE | ID: mdl-29108267

Down regulation of Protein Kinase D1 (PrKD1), a novel serine threonine kinase, in prostate, gastric, breast and colon cancers in humans leads to disease progression. While the down regulation of PrKD1 by DNA methylation in gastric cancer and by nuclear beta-catenin in colon cancer has been shown, the regulatory mechanisms in other cancers are unknown. Because we had demonstrated that PrKD1 is the only known kinase to phosphorylate threonine 120 (T120) of beta-catenin in prostate cancer resulting in increased nuclear beta-catenin, we explored the role of beta-catenin in gene regulation of PrKD1. An initial CHIP assay identified potential binding sites for beta-catenin in and downstream of PrKD1 promoter and sequencing confirmed recruitment of beta-catenin to a 166 base pairs sequence upstream of exon 2. Co-transfection studies with PrKD1-promoter-reporter suggested that beta-catenin represses PrKD1 promoter. Efforts to identify transcription factors that mediate the co-repressor effects of beta-catenin identified recruitment of both MYC and its obligate heterodimer MAX to the same binding site as beta-catenin on the PrKD1 promoter site. Moreover, treatment with MYC inhibitor rescued the co-repressor effect of beta-catenin on PrKD1 gene expression. Prostate specific knock out of PrKD1 in transgenic mice demonstrated increased nuclear expression of beta-catenin validating the in vitro studies. Functional studies showed that nuclear translocation of beta-catenin as a consequence of PrKD1 down regulation, increases AR transcriptional activity with attendant downstream effects on androgen responsive genes. In silico human gene expression analysis confirmed the down regulation of PrKD1 in metastatic prostate cancer correlated inversely with the expression of MAX, but not MYC, and positively with MXD1, a competing heterodimer of MAX, suggesting that the dimerization of MAX with either MYC or MXD1 regulates PrKD1 gene expression. The study has identified a novel auto-repressive loop that perpetuates PrKD1 down regulation through beta-catenin/MYC/MAX protein complex.

8.
Biomater Sci ; 5(12): 2437-2447, 2017 Nov 21.
Article En | MEDLINE | ID: mdl-29051963

Type-1 Diabetes (T1D) is a devastating autoimmune disorder which results in the destruction of beta cells within the pancreas. A promising treatment strategy for T1D is the replacement of the lost beta cell mass through implantation of immune-isolated microencapsulated islets referred to as the bioartificial pancreas. The goal of this approach is to restore blood glucose regulation and prevent the long-term comorbidities of T1D without the need for immunosuppressants. A major requirement in the quest to achieve this goal is to address the oxygen needs of islet cells. Islets are highly metabolically active and require a significant amount of oxygen for normal function. During the process of isolation, microencapsulation, and processing prior to transplantation, the islets' oxygen supply is disrupted, and a large amount of islet cells are therefore lost due to extended hypoxia, thus creating a major barrier to clinical success with this treatment. In this work, we have investigated the oxygen generating compounds, sodium percarbonate (SPO) and calcium peroxide (CPO) as potential supplemental oxygen sources for islets during isolation and encapsulation before and immediately after transplantation. First, SPO particles were used as an oxygen source for islets during isolation. Secondly, silicone films containing SPO were used to provide supplemental oxygen to islets for up to 4 days in culture. Finally, CPO was used as an oxygen source for encapsulated cells by co-encapsulating CPO particles with islets in permselective alginate microspheres. These studies provide an important proof of concept for the utilization of these oxygen generating materials to prevent beta cell death caused by hypoxia.


Bioartificial Organs , Diabetes Mellitus, Type 1/therapy , Insulin-Secreting Cells/transplantation , Oxygen/metabolism , Animals , Blood Glucose , Diabetes Mellitus, Type 1/pathology , Humans , Insulin-Secreting Cells/metabolism , Islets of Langerhans/metabolism , Islets of Langerhans Transplantation , Microspheres , Pancreas/metabolism , Pancreas/pathology , Pancreas Transplantation
9.
Methods Mol Biol ; 1479: 157-171, 2017.
Article En | MEDLINE | ID: mdl-27738934

Microencapsulation of islets is a procedure used to immunoisolate islets in order to obviate the need for immunosuppression of islet transplant recipients. Although microencapsulated islets have routinely been transplanted in the peritoneal cavity, the ideal site for their engraftment remains to be determined. The omentum, a highly vascularized tissue, has been proposed as an alternative site for microencapsulated islet transplantation. An added benefit to the omentum is that implanted microcapsules can be easily retrieved for post-transplant evaluation. This chapter describes a collagenase-based procedure for the retrieval of microencapsulated islets following the harvest of omentum pouch site of transplantation.


Cells, Immobilized/cytology , Diabetes Mellitus, Experimental/therapy , Islets of Langerhans Transplantation/methods , Islets of Langerhans/cytology , Omentum/surgery , Alginates/chemistry , Animals , Capsules/chemistry , Cell Survival , Cells, Immobilized/metabolism , Cells, Immobilized/transplantation , Drug Compounding , Graft Survival , Insulin/metabolism , Islets of Langerhans/metabolism , Mice , Rats
10.
Methods Mol Biol ; 1479: 225-235, 2017.
Article En | MEDLINE | ID: mdl-27738940

Two-dimensional (2D) culture systems do not represent the native microenvironment of the cells which is known to be three dimensional (3D), and surrounded by other cells from all directions. There exist interactions with other cell types in the same vicinity and this also cannot be replicated in a 2D culture. To study the cell-cell interactions between two or more cell types and their biological functions, a few 3D models have been used by different investigators. We have designed a 3D model to investigate the cell-cell interactions between various types of ovarian cells. The same model was also used to study the interactions between prostate cancer epithelial cells and stromal cells. This model uses hydrogel as the anchor matrix to fabricate the constructs and microencapsulation techniques to design multilayered microcapsules. In these multilayer microcapsules the different types of cells are compartmentalized by a sequential encapsulation process. In this chapter, we provide the protocol to compartmentalize two cell types in the same multilayer microcapsules. Although this chapter describes the fabrication of multilayer microcapsules with ovarian cells, the same approach could be applied to other multi-cell tissue-engineered constructs that require cell-cell interactions.


Alginates/chemistry , Cell Separation/methods , Coculture Techniques/methods , Ovary/cytology , Animals , Cells, Cultured , Drug Compounding/methods , Female , Glucuronic Acid/chemistry , Granulosa Cells/cytology , Hexuronic Acids/chemistry , Microscopy, Confocal/methods , Rats, Inbred F344 , Theca Cells/cytology , Tissue Engineering/methods
11.
Biomed Mater ; 11(6): 065009, 2016 11 11.
Article En | MEDLINE | ID: mdl-27834314

The in vivo function and phenotype of ovarian follicle cells are determined by many factors. When these cells are removed from the in vivo microenvironment and grown in a 2D in vitro environment, the function of the follicular cells is difficult to preserve. A collagen hydrogel was used to examine the hormone and oocyte maturation of ovary follicles in a 3D culture system. Ovarian follicles from rats were isolated and cultured in various concentration of type I collagen hydrogels ranging from 1% to 7% (weight/volume). Differences in cell survival, follicle growth and development, sex hormone production, and oocyte maturation were seen with the modifications in the collagen hydrogel density and elasticity. The results show the significance of the collagen hydrogel properties on phenotype and function maintenance of the ovarian follicles in a 3D culture system.


Cell Culture Techniques/methods , Collagen/chemistry , Hydrogels/chemistry , Ovarian Follicle/physiology , Animals , Cell Survival , Cells, Cultured , Elasticity , Estradiol/chemistry , Female , Humans , Meiosis , Oocytes/cytology , Phenotype , Progesterone/chemistry , Rats , Rats, Inbred F344 , Rheology , Viscosity
12.
Ann Surg ; 264(1): 169-79, 2016 07.
Article En | MEDLINE | ID: mdl-26649588

OBJECTIVES: Our study aims at producing acellular extracellular matrix scaffolds from the human pancreas (hpaECMs) as a first critical step toward the production of a new-generation, fully human-derived bioartificial endocrine pancreas. In this bioartificial endocrine pancreas, the hardware will be represented by hpaECMs, whereas the software will consist in the cellular compartment generated from patient's own cells. BACKGROUND: Extracellular matrix (ECM)-based scaffolds obtained through the decellularization of native organs have become the favored platform in the field of complex organ bioengineering. However, the paradigm is now switching from the porcine to the human model. METHODS: To achieve our goal, human pancreata were decellularized with Triton-based solution and thoroughly characterized. Primary endpoints were complete cell and DNA clearance, preservation of ECM components, growth factors and stiffness, ability to induce angiogenesis, conservation of the framework of the innate vasculature, and immunogenicity. Secondary endpoint was hpaECMs' ability to sustain growth and function of human islet and human primary pancreatic endothelial cells. RESULTS: Results show that hpaECMs can be successfully and consistently produced from human pancreata and maintain their innate molecular and spatial framework and stiffness, and vital growth factors. Importantly, hpaECMs inhibit human naïve CD4 T-cell expansion in response to polyclonal stimuli by inducing their apoptosis and promoting their conversion into regulatory T cells. hpaECMs are cytocompatible and supportive of representative pancreatic cell types. DISCUSSION: We, therefore, conclude that hpaECMs has the potential to become an ideal platform for investigations aiming at the manufacturing of a regenerative medicine-inspired bioartificial endocrine pancreas.


Extracellular Matrix/metabolism , Pancreas , Tissue Engineering , Tissue Scaffolds , Humans , Islets of Langerhans/metabolism , Organogenesis , Pancreas/metabolism , Regeneration , Tissue Engineering/methods
13.
Pancreas ; 43(4): 605-13, 2014 May.
Article En | MEDLINE | ID: mdl-24681880

OBJECTIVE: Our study aim was to determine encapsulated islet graft viability in an omentum pouch and the effect of fibroblast growth factor 1 (FGF-1) released from our redesigned alginate microcapsules on the function of the graft. METHODS: Isolated rat islets were encapsulated in an inner core made with 1.5% low-viscosity-high-mannuronic-acid alginate followed by an external layer made with 1.25% low-viscosity high-guluronic acid alginate with or without FGF-1, in microcapsules measuring 300 to 400 µm in diameter. The 2 alginate layers were separated by a perm-selective membrane made with 0.1% poly-L-ornithine, and the inner low-viscosity-high-mannuronic-acid core was partially chelated using 55 mM sodium citrate for 2 minutes. RESULTS: A marginal mass of encapsulated islet allografts (∼2000 islets/kg) in streptozotocin-diabetic Lewis rats caused significant reduction in blood glucose levels similar to the effect observed with encapsulated islet isografts. Transplantation of alloislets coencapsulated with FGF-1 did not result in better glycemic control, but induced greater body weight maintenance in transplant recipients compared with those that received only alloislets. Histological examination of the retrieved tissue demonstrated morphologically and functionally intact islets in the microcapsules, with no signs of fibrosis. CONCLUSIONS: We conclude that the omentum is a viable site for encapsulated islet transplantation.


Alginates , Diabetes Mellitus, Experimental/surgery , Graft Survival , Immunocompetence , Islets of Langerhans Transplantation/methods , Islets of Langerhans/metabolism , Omentum/surgery , Tissue Scaffolds , Angiogenesis Inducing Agents/administration & dosage , Animals , Blood Glucose/metabolism , Cell Survival , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/immunology , Fibroblast Growth Factor 1/administration & dosage , Glucuronic Acid , Hexuronic Acids , Islets of Langerhans/blood supply , Islets of Langerhans/immunology , Male , Neovascularization, Physiologic , Omentum/blood supply , Rats, Inbred Lew , Rats, Wistar , Time Factors
14.
J Urol ; 191(3): 850-9, 2014 Mar.
Article En | MEDLINE | ID: mdl-23973520

PURPOSE: Cellular therapy induced transient urodynamic improvement in a rat model of Parkinson disease in which bladder dysfunction was noted after unilateral injection of 6-hydroxydopamine into the medial forebrain bundle. We sought to prolong the effect by injecting allogeneic rat bone marrow mesenchymal stromal cells before and after microencapsulation into the substantia nigra pars compacta. MATERIALS AND METHODS: Female rats underwent unilateral stereotactic injection of 6-hydroxydopamine in the medial forebrain bundle. Injection was performed in the ipsilateral substantia nigra pars compacta using vehicle alone or vehicle with nonmicroencapsulated or microencapsulated rat bone marrow derived mesenchymal stromal cells. Rats were evaluated by cystometry 7, 14, 28 and 42 days after treatment. Brains were extracted for immunostaining. RESULTS: At 42 days the nonmicroencapsulated group had lower threshold and intermicturition pressure, spontaneous activity and AUC than vehicle treated animals. Rats that received microencapsulated cells had lower threshold pressure at 28 days and lower spontaneous activity at 42 days than vehicle treated rats. Microencapsulated and nonmicroencapsulated rat bone marrow derived mesenchymal stromal cells were noted in the substantia nigra pars compacta up to 42 days after transplantation. At 42 days tyrosine hydroxylase positive neurons were more numerous in the substantia nigra pars compacta of the nonmicroencapsulated group, followed by the microencapsulated and vehicle treated groups. CONCLUSIONS: Urodynamic effects of the 6-hydroxydopamine lesion persisted up to 42 days after vehicle injection. Transplantation of nonmicroencapsulated rat bone marrow derived mesenchymal stromal cells improved urodynamic pressure by 42 days after treatment more markedly than microencapsulated cells. This was associated with more tyrosine hydroxylase positive neurons in the treated substantia nigra pars compacta of the nonmicroencapsulated group, suggesting that functional improvement requires a juxtacrine effect.


Bone Marrow/physiopathology , Cell- and Tissue-Based Therapy , Mesenchymal Stem Cell Transplantation , Parkinson Disease/physiopathology , Parkinson Disease/therapy , Urodynamics , Animals , Disease Models, Animal , Female , Flow Cytometry , Immunohistochemistry , Microscopy, Confocal , Oxidopamine , Rats , Rats, Sprague-Dawley , Substantia Nigra/physiopathology
15.
Maturitas ; 77(1): 12-9, 2014 Jan.
Article En | MEDLINE | ID: mdl-24210634

The goal of regenerative medicine is to repair, replace, or regenerate diseased tissues/organs in order to restore normal function. In this paper we will first discuss the general principle of regenerative medicine and the various techniques and approaches that have been used to replace or regenerate cells in diseased tissues and organs. Then, we will review different regenerative medicine approaches that have been used to treat specific diseased tissues and organs of the reproductive system in both animal and human experiments. It is clear from this article that regenerative medicine holds significant promise, and we hope that the review will serve as a platform for further development of regenerative medicine technologies for the treatment of inadequacies of the reproductive system.


Female Urogenital Diseases/therapy , Regenerative Medicine , Tissue Engineering , Urogenital System , Animals , Female , Humans
16.
PLoS One ; 8(9): e75187, 2013.
Article En | MEDLINE | ID: mdl-24073251

Paracrine function is a major mechanism of cell-cell communication within tissue microenvironment in normal development and disease. In vitro cell culture models simulating tissue or tumor microenvironment are necessary tools to delineate epithelial-stromal interactions including paracrine function, yet an ideal three-dimensional (3D) tumor model specifically studying paracrine function is currently lacking. In order to fill this void we developed a novel 3D co-culture model in double-layered alginate hydrogel microspheres, incorporating prostate cancer epithelial and stromal cells in separate compartments of the microspheres. The cells remained confined and viable within their respective spheres for over 30 days. As a proof of principle regarding paracrine function of the model, we measured shedded component of E-cadherin (sE-cad) in the conditioned media, a major membrane bound cell adhesive molecule that is highly dysregulated in cancers including prostate cancer. In addition to demonstrating that sE-cad can be reliably quantified in the conditioned media, the time course experiments also demonstrated that the amount of sE-cad is influenced by epithelial-stromal interaction. In conclusion, the study establishes a novel 3D in vitro co-culture model that can be used to study cell-cell paracrine interaction.


Cell Communication , Epithelial Cells/pathology , Prostatic Neoplasms/pathology , Stromal Cells/pathology , Cadherins/metabolism , Cell Survival , Coculture Techniques , Culture Media, Conditioned/pharmacology , Enzyme-Linked Immunosorbent Assay , Epithelial Cells/metabolism , Humans , Male , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Paracrine Communication , Prostatic Neoplasms/metabolism , Stromal Cells/metabolism , Tumor Cells, Cultured , Tumor Microenvironment
17.
Biomaterials ; 34(10): 2412-20, 2013 Mar.
Article En | MEDLINE | ID: mdl-23274068

Although hormone replacement therapy is an option for the loss of ovarian function, hormone delivery through pharmacological means results in various clinical complications. The present study was designed to deliver sex steroids by a functional construct fabricated using encapsulation techniques. Theca and granulosa cells isolated from ovaries of 21-day old rats were encapsulated in multilayer alginate microcapsules to recapitulate the native follicular structure. Cells encapsulated in two other schemes were used as controls to assess the importance of the multilayer structure. The endocrine functions of the encapsulated cells were assessed in vitro for a period of 30 days. Encapsulated cells showed sustained viability during long-term in vitro culture with those encapsulated in multilayer capsules secreting significantly higher and sustained concentrations of 17 ß-estradiol (E(2)) than the two other encapsulation schemes (p < 0.05, n = 6) in response to follicle-stimulating hormone (FSH) and luteinizing hormone (LH). In addition, cells in the multilayer microcapsules also secreted activin and inhibin in vitro. In contrast, when granulosa and theca cells were cultured in 2D culture, progesterone (P(4)) secretion increased while E(2) secretion decreased over a 30-day period. In summary, we have designed a multilayer engineered ovarian tissue that secretes sex steroids and peptide hormones and responds to gonadotropins, thus demonstrating the ability to recapitulate native ovarian structure ex vivo.


Gonadotropins/pharmacology , Ovary/cytology , Ovary/metabolism , Peptide Hormones/metabolism , Steroids/metabolism , Animals , Cells, Cultured , Endocrine Cells/drug effects , Female , Flow Cytometry , Ovary/drug effects , Rats , Rats, Inbred F344 , Tissue Engineering
18.
Expert Opin Biol Ther ; 13(1): 103-13, 2013 Jan.
Article En | MEDLINE | ID: mdl-23110384

INTRODUCTION: The most critical issue to organ transplantation is the identification of new sources of organs. The present manuscript illustrates the state-of-the-art regenerative medicine (RM) investigations aiming to manufacturing abdominal organs for transplant purposes. AREAS COVERED: This manuscript focuses on research in the bioengineering and regeneration of kidneys, insulin-producing cells, livers and small bowel. The main technology currently under development exploits the seeding of cells on supporting scaffolding material. Despite favorable preliminary results obtained with relatively simple, hollow organs, when more complex organs are considered, the scenario changes dramatically. Investigations are still in early stages, and clinical translation is not yet foreseeable based on current knowledge and information. Obstacles are numerous but we believe the critical factor hampering success is lack of in-depth understanding of the extracellular matrix (ECM) and cell-ECM interactions, as well as the mechanisms with which organs develop in utero. EXPERT OPINION: The success of RM to generate transplantable abdominal organs relies heavily on progress in (stem) cell therapies, developmental and ECM biology, and in the thorough understanding of the intricate relationship and interplay between cells and the ECM. This will require enormous investments in financial and medical resources, which ideally should be embarked upon by governments, the private sector and academia.


Abdomen , Bioengineering , Organ Transplantation , Regeneration , Humans
19.
Clin Chim Acta ; 414: 281-8, 2012 Dec 24.
Article En | MEDLINE | ID: mdl-23041218

BACKGROUND: Insulin-like growth factor (IGF) system components are important regulators of bone metabolism, which have a predominant role in determining bone mineral density (BMD). While the serum levels of IGF-I are regulated by various systemic hormones and growth factors, IGF-II levels reflect the skeletal production relative to physical activity, mechanical loading, aging, race etc. Though various studies have been carried out among women of different ethnic groups to understand the relationship between serum levels of IGF-II and BMD, the results seem to be quite inconclusive. METHODS: We evaluated the same, recruiting South-Indian women who engage themselves in a wide variety of physical activities pertaining to their profession and life style. RESULTS: Serum levels of IGF-II and IGF binding protein (IGFBP)-3 showed positive correlation with calcaneal BMD, whereas IGFBP-4 showed negative correlation. These IGF system components exhibited similar correlations with serum bone formation markers and opposite trend with bone resorption marker. While both IGF-II and IGFBP-3 levels were observed to be decreased with aging and menopause, IGFBP-4 levels increased. CONCLUSIONS: The alterations in serum levels of IGF-II and its binding proteins due to aging and menopause could be some of the major contributors of decreased calcaneal BMD observed among elderly women.


Aging/blood , Bone Density , Calcaneus/chemistry , Insulin-Like Growth Factor Binding Proteins/blood , Insulin-Like Growth Factor II/analysis , Postmenopause/blood , Adult , Aged , Aged, 80 and over , Cross-Sectional Studies , Female , Humans , India , Insulin-Like Growth Factor Binding Protein 3/blood , Insulin-Like Growth Factor Binding Protein 4/blood , Middle Aged , Reference Values , Young Adult
20.
Integr Cancer Ther ; 10(4): 328-40, 2011 Dec.
Article En | MEDLINE | ID: mdl-21196432

Melanoma is an aggressive tumor that expresses the pigmentation enzyme tyrosinase. Tyrosinase expression increases during tumorigenesis, which could allow for selective treatment of this tumor type by strategies that use tyrosinase activity. Approaches targeting tyrosinase would involve gene transcription or signal transduction pathways mediated by p53 in a direct or indirect manner. Two pathways are proposed for exploiting tyrosinase expression: (a) a p53-dependent pathway leading to apoptosis or arrest and (b) a reactive oxygen species-mediated induction of endoplasmic reticulum stress in p53 mutant tumors. Both strategies could use tyrosinase-mediated activation of quercetin, a dietary polyphenol that induces the expression of p53 and modulates reactive oxygen species. In addition to antitumor signaling properties, activation of quercetin could complement conventional cancer therapy by the induction of phase II detoxification enzymes resulting in p53 stabilization and transduction of its downstream targets. In conclusion, recent advances in tyrosinase enzymology, prodrug chemistry, and modern chemotherapeutics present an intriguing and selective multitherapy targeting system where dietary bioflavonoids could be used to complement conventional cancer treatments.


Genes, p53 , Melanoma/enzymology , Monophenol Monooxygenase/metabolism , Neuroblastoma/enzymology , Quercetin/pharmacology , Skin Neoplasms/enzymology , Animals , Apoptosis , Genes, p53/drug effects , Humans , Melanoma/drug therapy , Melanoma/genetics , Monophenol Monooxygenase/genetics , Neuroblastoma/genetics , Quercetin/metabolism , Quercetin/pharmacokinetics , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Skin Neoplasms/drug therapy , Skin Neoplasms/genetics
...